Skip to main content

Association of immune cell recruitment and BPD development

Abstract

In the neonatal lung, exposure to both prenatal and early postnatal risk factors converge into the development of injury and ultimately chronic disease, also known as bronchopulmonary dysplasia (BPD). The focus of many studies has been the characteristic inflammatory responses provoked by these exposures. Here, we review the relationship between immaturity and prenatal conditions, as well as postnatal exposure to mechanical ventilation and oxygen toxicity, with the imbalance of pro- and anti-inflammatory regulatory networks. In these conditions, cytokine release, protease activity, and sustained presence of innate immune cells in the lung result in pathologic processes contributing to lung injury. We highlight the recruitment and function of myeloid innate immune cells, in particular, neutrophils and monocyte/macrophages in the BPD lung in human patients and animal models. We also discuss dissimilarities between the infant and adult immune system as a basis for the development of novel therapeutic strategies.

Background

Bronchopulmonary dysplasia (BPD) is the neonatal form of chronic lung disease occurring in the context of prematurity and is characterized by impaired pulmonary development [1]. The consequences of misdirected lung development persist into adulthood, and although the advances in neonatal intensive care decreased the rate of overall mortality after premature birth, the prevalence of chronic complications like BPD remained [2, 3]. Different prenatal and postnatal factors have been introduced as contributors to BPD development, including genetic/epigenetic risk factors, intrauterine hypoxia and growth retardation, infection, mechanical ventilation (MV), and oxygen supplementation [4, 5]. Clinically, BPD is classified into three severity grades according to the need for oxygen supplementation or MV 28 days after birth and near term age, i.e., 36 weeks gestational age (GA) [6]. More than half of the preterm infants that require life-saving postnatal treatment with MV or oxygen supplementation develop subsequent complications such as failure of alveolo- and angiogenesis [7, 8]. Premature babies born before 32 weeks of GA present with a structural and functional immaturity of the lung that includes an immune system not yet equipped to sufficiently respond to environmental insults. The subsequent vulnerability to infections and injury together with the misdirected role of immune functions in organ development highlights the importance of better understanding—and potentially targeting—immune-related phenomena [9]. Inflammation is a vital element of host defense [10]. Excessive or persistent inflammation, however, is known to interfere with organ development including the lung, and thus, a key contributor to the emergence and pathogenesis of BPD indicating disease progression [11, 12]. Inflammation can be triggered by both prenatal as well as postnatal factors including hyperoxia, MV, and infections [13,14,15,16] that act beyond the background, e.g., immaturity and gender [16]. Preterm labor per se is associated with intrauterine infections with ureaplasma and mycoplasma infections holding a specific role in BPD development [13]. In consequence, intrauterine and early postnatal infections and the respective inflammation are common in premature infants [17].. The immune system in premature infants is still undeveloped with a lower number of neutrophils and monocytes in the cord blood, resulting in a greater vulnerability to infections and a significant imbalance of pro-and anti-inflammatory mechanisms upon injury [18, 19].

The later development of BPD is then characterized by the accumulation and activation of myeloid leukocytes cells in the lung, which in turn drive pathophysiological processes such as an enhanced permeability of the endothelial and epithelial barrier (“leakage”) [20, 21]. Recently, studies specifically focused on the impact of the accumulation of inflammatory cells, myeloid neutrophils, and monocytes in particular, on misguided alveolar and pulmonary vascular development as the hallmark of BPD [22,23,24] However, the bidirectional role of resident and recruited myeloid cells in development and injury has been targeted for the development of treatment strategies. This review provides an overview of the current state of knowledge available on the known and potential role of neutrophils, monocyte/macrophages, and their crosstalk with resident cells in BPD development, side by side with a discussion about the main challenges concerning modeling the disease and future perspectives in the field. The important differences in immune functions when compared to adults are highlighted.

Innate immunity in the development of neonatal chronic lung disease

Due to the limited exposure to foreign antigens in utero, the newborn infant relies on innate immunity-dependent defense strategies as the adaptive immune response is still naïve [25,26,27]. Also, the shortened time for the prenatal development of immune functions in preterm infants impacts immune responses immediately after birth [28]. Circulating and resident myeloid immune cells such as neutrophils and monocytes are at the forefront of the innate immune response and act as potential triggers of inflammatory signals including cytokines and chemokines [29], which mediate immune activation as well as the transition from innate to adaptive immunity [30, 31].

Environmental assaults not only encompass bacterial or viral infections but exposure to toxins and mechanical stress as initiated by MV and oxygen supplementation all triggering inflammation involving both innate and adaptive immune responses [32, 33] (see Fig. 1). The airway epithelium is an immunologically active barrier and the main source of pro-inflammatory cytokines [34, 35]. Upon the airway epithelial injury and local release of the classical pro-inflammatory cytokines of the innate immune defense such as interleukin-1, -6, -8 (IL-1, IL-6, IL-8), and tumor necrosis factor-alpha (TNFα), blood neutrophils immediately migrate into the lung tissue, subsequently followed by monocytes which—once recruited—rapidly differentiate into macrophages [36,37,38]. This monocyte-to-macrophage differentiation is initiated after the emigration from the circulation into the tissue in association with gaining different functional phenotypes dependent on the local tissue environment [39]. Depending on the micro-environmental signals, macrophages develop distinct functions represented in the concept of their differentiation into classical activated, inflammatory (M1) or activated, anti-inflammatory, or fibrotic (M2) states [38]. Furthermore, neonatal and adult alveolar macrophages (AMs) differ from each other and present their unique transcriptome profile under the impact of micro-environmental signals [40].

Fig. 1
figure 1

Schematic represents the innate immune signals related to lung inflammation culminating in the BPD development and progression. Preterm infants suffer from BPD due to the impact of various risk factors including genetic background, prenatal and postnatal infections, nutrition, oxygen toxicity, and mechanical ventilation. Exposure of the structurally and functionally immature lung to these risk factors provokes oxidative stress and results in the increased expression of pro-inflammatory cytokines by resident cells in the alveolar niche. Subsequently, innate immune cells are recruited including neutrophils as the first-line defense. These events are followed by the extravasation of monocytes which eventually differentiate into macrophages in the tissue context. Neutrophil and monocyte signaling is associated with pulmonary tissue damage including impairment of epithelial and vascular function and progression of inflammatory processes. Black arrows indicate the elevating events during BPD

Studies on BPD research during the last few decades primarily focused on the association between cytokine and chemokine expression patterns and disease onset or progress [41]. Researchers found high expression levels of specific cytokines stemming from classical macrophage activation such as monocyte chemoattractant protein-1 (MCP-1/CCL2), macrophage inflammatory proteins (MIP), the neutrophil chemokine IL-8, and low expression of IL-10 in association with BPD development [12, 42,43,44,45] while expression other cytokines, for example, IL-4 and IL-13 did not correlate with BPD [46]. In addition to the myeloid lineage, an increase in type 3 innate lymphoid cells (ILC3) have been reported for their pro-inflammatory role in BPD development, mainly through the secretion of IL-17, which is known as a key factor in the recruitment of neutrophils via stimulation of IL-8 including neutrophils and CXCL1 and CXCL2 chemokines [47, 48]. In contrast, levels of granulocyte colony-stimulating factor (G-CSF), responsible for the stimulation of granulocyte production in the bone marrow (BM), were found to be reduced in preterm when compared to term infants, which indicates the defective function of neutrophils during infection [49]. Thus, a potential therapeutic approach to treat or prevent BPD has been proposed by inhibiting the unsolicited expression of cytokine and chemokine [50, 51], which plays a critical role in preterm infants innate immunity and BPD development.

To delineate specific cellular functions engaged in the innate immune response, we, in the following, specifically outline neutrophil and monocyte/macrophage functions and interactions in light of the sequential order of immune events.

Neonatal neutrophils and their activation in BPD

Neutrophils play an indispensable role in acute lung inflammation in both mature and developing organisms [52,53,54]. During the initiation phase of inflammation, neutrophils undergo a variety of changes in gene expression and functional properties [55]. Neutrophilic granulocytes originate in the BM and are released into circulation [56], where they act as the first line of cellular immune defense when getting recruited to the site of injury [57]. This first wave of inflammation is followed by the recruitment of monocytes in a later stage. Neutrophils eliminate pathogens through phagocytosis and via releasing proteases, reactive oxygen species (ROS), and bioactive membrane vesicles through a function called degranulation [58]. ROS are highly reactive species formed from O2, such as hydroperoxyl and hydrogen peroxide, and can be transformed into radicals [57, 59]. The sudden postnatal exposure of the immature lung to oxygen and MV is currently seen as the inducer of ROS-dependent local and systemic neutrophilic inflammatory responses [60,61,62], that in turn can cause the release and activation of neutrophils and increased ROS production [63, 64]. Next to the ROS-induced effects that restrict lung development and contribute to BPD, another destructive role of activated neutrophils in the process of alveolar formation is likely related to the release of exosomes that can increase the proliferation of airway smooth muscle cells, induce remodeling and extracellular matrix (ECM) destruction—all being features of BPD [65,66,67,68]. In addition, the release of neutrophil elastase (NE) and metalloproteinases (MMP) like MMP-9 can cleave fibronectin and increase ECM degradation in the alveoli, thereby contributing to scaffold remodeling and BPD progress [69, 70]. Alternatively, these factors can be released by the ECM itself, thereby perpetuating the vicious cycle of damaging the alveolar niche [68]. In line with this, endogenous protease inhibitors, such as serine protease inhibitor (SERPIN)B1 have been demonstrated to hold protective functions against neutrophil protease-induced tissue damage in BPD [71].

In comparison to adult immune functions, term and preterm newborn infants show fewer peripheral blood neutrophil counts [72]. At 22–23 weeks of gestation, only 2% of all leukocytes in the peripheral blood are neutrophils, while these counts increase to 60% in term-born infants [55] and about 60–70% in adults [73]. Functionally, neonatal neutrophils show the deficiency to form neutrophil extracellular traps (NETs) [74] as well as a lower cell surface expression of L-selectin [72]. Following injury, endothelial cells express leucocyte adhesion molecules on their luminal side, particularly P- and E-selectins, together with several integrin members of the ICAM family, to allow adhesion of neutrophils through their respective selectin ligands and integrins, e.g., CD18 and CD11b [75]. Neonatal neutrophils, however, express lower levels of CD11b and subsequently display only attenuated abilities for adhesion as compared to adult cells. Also, reduced neutrophil CD18 and L-Selectin expression levels have even been described for their predictive value in BPD [76] while it has been reported that the ratio of blood neutrophil to lymphocyte has been increased 72 h after birth [12]. The latter points to an inflammatory stimulation in utero encompassing the lung. Nevertheless, reduced blood neutrophil levels have to be viewed in light of a potential imbalance of tissue accumulation and BM production [77]. The vulnerability of the newborn lung towards inflammatory injury is highlighted by findings in bronchoalveolar lavage fluid (BALF) demonstrating the combination of MV, hyperoxia, and inflammation to trigger the expression of acute-phase cytokines and chemokines in newborn infants [78] together with increased neutrophil apoptosis where intact neutrophils are phagocytosed by AMs before degranulation [79]. These findings might reflect a mechanism contributing to sustained inflammation and tissue damage. Neutrophil-related events as a therapeutic approach are still being explored.

The second wave of inflammation, induced by the neutrophil through releasing chemoattractants, involves the recruitment of other immune cells, specifically monocytes/macrophages.

Monocytes/macrophages and their response in BPD developing infants

Since the lung is the interface to the external environment and related immunological challenges [80], mononuclear phagocytic cells are dedicated to confronting these challenges. Therefore, in response to environmental insults, circulating monocytes migrate into the alveolar space and contribute to the process of alveolar injury and remodeling [81, 82]. Monocyte/monocyte-derived alveolar macrophages (Mo-AMs) recruitment to the lung is a characteristic process in BPD [83], even in light of a reduced number of monocytes in the neonatal peripheral tissue per se [84] and their lower adherence and transmigration capacity when facing the non-inflamed endothelium [84].

The process seems to mark chronification in BPD despite the potential to contribute to the resolution of inflammation in the first place. Like neutrophils, monocytes originate from the granulocyte monocyte progenitor (GMP) in the BM and play a key role in innate immune responses which are closely related to vascular homeostasis [85, 86] and contribute to processes such as phagocytosis and lung regeneration at the same time. These functions have in part been attributed to modulating alveolar stem cells [87, 88].

Current concepts relate to three human monocyte subsets: classical (CD14+CD16), non-classical (CD14-CD16+), and intermediate (CD14+CD16+) monocytes [89]. Each subset holds unique functions represented by different surface markers [90]. The differing abilities regarding recruitment, cytokine production, and capability of endothelial activation [91] relate to the grouping introduced above. Whereas nonclassical/intermediate monocytes secrete higher levels of TNFα and demonstrate an increased capacity for endothelial transmigration, classical monocytes secrete high levels of IL-6, and assist neutrophil recruitment [91]. In mice—in relation to the human concept—main types of monocytes are identified according to three subsets including classical (Ly6C++, CCR2++, CD43+), nonclassical (Ly6C+, CCR2+, CD43++, CX3CR1+), and intermediate (Ly6C++,Treml4+, CD43++) monocytes [92,93,94,95]. Here, Ly6C+ nonclassical monocytes continuously scan the endothelium for injury and infections [96] and are thereby involved in maintaining endothelial barrier integrity, regeneration, and repair [97,98,99].

Monocytes respond to chemokine signals with the adhesion to endothelial cells following a multistep adhesion cascade involving the interaction of leukocyte adhesion molecules for instance L-selectin, PSGL1, LFA1, MAC1, VLA4, and their respective receptors on endothelial cells from the selectin and immunoglobulin superfamily [85, 86, 100,101,102]. Adhesion dynamics and homeostatic extravasation is depending on CD31 and CD54 expression levels [84]. Monocytes from preterm infants, however, show a lower surface expression of the CD11b/CD18 and CD31 adhesion receptors, resulting in a reduced ability for extravasation and greater susceptibility to infections [84, 103]. Necessary for the subsequent differentiation of fetal monocytes into pre-AMs during embryonic development, the transforming growth factor (TGF)-β1 is simultaneously a key player in lung development. Its dual role next in inflammation and apoptosis [104, 105] and the dynamic regulation during alveolarization renders the prenatal regulation of TGF-β during in utero inflammation an interesting target [106, 107]. TGF-β1 is an important factor for AM maturation after birth as well as for the homeostasis of adult AMs [108] and provides important signals for monocyte recruitment [109] and their activation. Whereas CD14+ monocytes stimulate the TGF-β1 pathway through the expression of the integrin αvβ8, this is not observed for CD16+ monocytes [110].

After exiting the circulation, blood-derived monocytes rapidly differentiate into highly phagocytic active macrophages which are innate immune cells that are abundant in tissues [111]. In many organs, including the lung, they are divided into tissue-resident macrophages (TRMs) and “monocyte-derived” macrophages (Mo-Ms) [38, 82, 111]. While the latter are continuously generated and replaced from the BM hematopoietic system, TRMs are derived during embryonic development from erythro-myeloid progenitors of the yolk sac (YS) and fetal liver monocytes [112, 113]. TRMs are abundant in lung alveoli and contribute to the formation of the alveolar niche in the first week of life [114]. TRMs can develop from fetal monocytes by gaining an established phenotype shortly after birth in response to instructive cytokines. Furthermore, they are able to self-maintain throughout life [115]. YS-derived macrophages and fetal monocytes can arise as identical alveolar macrophages, while mostly the fetal monocytes colonize the alveolar area [116]. Tissue-resident alveolar macrophages (TRM-AM) play a central role in lung development, tissue homeostasis, and immune responses. Their absence leads to infections and alveolar proteinosis due to loss of protein clearance [115]. Their contribution to the formation of the alveolar niche in the first weeks of life as well as to disease development is of current interest [112,113,114,115, 117, 118]. Although the exact contribution of the macrophage lung in the pathogenesis of BPD has not been elucidated yet, adequate early macrophage activation of macrophages is shown to be crucial to protecting infants lungs from the development of BPD [119].

Several studies have shown that Mo-AMs play an important role in endotoxin-induced, acute lung inflammation [52, 120,121,122]. However, as the maturation of the lung macrophages is a postnatal process, preterm and term newborns are rendered more susceptible to disease until a “catch up” is achieved if at all [123], further aggravated by the reduced number of lung TRM-AM [124]. Monocytes and Mo-AMs from preterm infants less than 30 weeks of gestation, even show impaired pro-inflammatory cytokine production, i.e., IL-8, IL-1β, IL-6, and TNFα [9, 125,126,127], together with a deficient pathogen response in comparison to adults, enhancing the infection and damage susceptibility further [128].

Both MV and oxygen exposure of the underdeveloped lung provokes oxidative stress [63, 129], in turn leading to increased expression of pro-inflammatory cytokines such as TNFα, IL-8, IL-1β, and IL-6 tracheobronchial aspirates [130,131,132]. These multiple-hit events are self-perpetuating when adding the crosstalk with the epithelial cells engaged in cytokine expression, NF-κB activity, and ROS production which results in monocyte recruitment and macrophage differentiation [133].

Disease modeling of BPD by focusing on innate immune cells

The role of neutrophils and monocytes in BPD onset and progression is studied using different methods and models, mainly relying on 2D cell cultures and measurement in clinical samples to complex animal models (Table 1). Several studies addressed the innate immune system in the lung compartment by the use of BAL, TA specimens, cord blood cells, and serum from preterm and term neonates in the first week of life [24, 45, 134,135,136]. Patient-derived samples are valuable pre-clinical tools for defining predictive biomarkers in disease development. However, isolating the untouched innate immune cells for in vitro studies can significantly affect their phenotype [137]. Several non-human species studies, which used mice, sheep, and baboons have been performed to study the BPD [13, 71, 124]. The experimental use of both prenatal and postnatal lung injury including MV and oxygen exposure in wild-type and transgenic animals attempts to investigate the cellular and molecular mechanisms of the development of disease in a preclinical setting [8, 13, 124, 138]. Differences between animal and human development of immune functions render these studies challenging. In addition, different in vitro models using primary cells or primary cell lines are added to gain insight into injury-relevant mechanisms of the innate immune cell [49, 139]. However, to the best of our knowledge, realistic in vitro models which mimic the physiological and pathophysiological of the lung with a focus on the migration of neutrophils and monocytes under static or dynamic conditions have not been employed in the field of BPD research thus far [139,140,141].

To provide an overview, we summarized relevant models for studying neutrophil and monocyte functions in the injured developing lung in chronological order in Table 1.

Table 1 Selected research on the role of neutrophils and monocytes in BPD

Conclusions and outlook

In summary, the immature lung undergoing injury is characterized by increased concentrations of proinflammatory cytokines as exemplified by measurements in TA obtained from very premature infants in the first days of life. While infections, oxygen exposure, and MV contribute to BPD development, the affected innate immune functions play a critical role in this complex disease of prematurity. During the past few years, significant efforts have been made to examine the role of inflammation-induced lung injury considering both prenatal and postnatal conditions while using human samples and animal models. Studies showed that a preterm neonate with a structurally and functionally immature lung, paired with an underdeveloped immune response exhibit an increased pro-inflammatory response when BPD develops. Therefore, targeting the inflammatory process is considered a therapeutic or preventive approach for BPD.

Nonetheless, many conflicting results remain. Prematurity as such results in a smaller pool of neutrophils and monocyte/macrophages and a subsequently lower capacity to secrete cytokines and reach pathogen clearance. In BPD, however, elevated numbers of neutrophils and monocytes, and their inflammatory cytokines can be detected in the diseased lung. The differences in local and systemic concentrations may relate to mechanisms driving tissue accumulation despite reduced BM production rates. Targeting the number of peripheral blood immune cells has been suggested as a therapeutic strategy in BPD. Nevertheless, this approach neglects the developmental functions, the same innate immune cells, and their signaling molecules hold.

Concerning cell-specific effects and their interplay, neutrophil presence and activation marking the first line of defense hold a crucial function in the BPD lung and correlate with the severity of symptoms. Detection and characterization of a pronounced monocyte-centered inflammatory response in preterm infants with later BPD mark the “second wave” of events. However, the challenging question is how physiologic defense signaling is changed into an immune reaction while infection clearing and termination of inflammation are not reached. The subsequent release of damage signals initiates the detrimental cascade of alveolar and airway injury.

Future studies need to overcome current limitations that exist due to a variety of reasons. First of all—given the nature of the patient collective—only a limited number of preterm infants can be enrolled in the majority of studies. Secondly, volume restrictions of clinical samples obtained from preterm infants limit the experimental settings in addition to the non-physiological activation of primary cells during isolation and culture. Hence, reliable analysis techniques and the avoidance of solely targeted approaches need to be combined with optimized “preclinical” in vitro models to overcome the described challenges. The establishment of large datasets standardized or harmonized between centers and groups including single-cell analysis will help to provide valuable resources for studies to come regarding both developmental and injury functions of the innate immune system. Preclinical animal models—despite the differences in immune regulation processes and marker expression when comparing mice and men—should be combined to tackle context-related knowledge gaps putting in vitro findings into perspective concerning cellular crosstalk and spatial resolution.

In the future, our understanding of the role of the immune system in BPD will become increasingly important as more sophisticated therapeutic and diagnostic possibilities emerge that may allow targeted approaches to treat and monitor disease-relevant processes in a population tackling critical steps of organ development.

Availability of data and materials

Not applicable.

Abbreviations

MV:

Mechanical ventilation

GA:

Gestational age

BAL:

Bronchoalveolar lavage

TA:

Tracheal aspirate

bpd:

Bronchopulmonary dysplasia

nCLD:

Neonatal chronic lung disease

AMs:

Alveolar macrophages

TRMs:

Tissue-resident macrophages

Mo-Ms:

Monocyte-derived” macrophages

TR-AMs:

Tissue-resident alveolar macrophages

Mo-AMs:

Monocyte-derived alveolar macrophages

RDS:

Respiratory distress syndrome

BLF:

Bronchoalveolar lavage fluid

NETs:

Neutrophil extracellular traps

TGF-β1:

Transforming growth factor

G-CSF:

Granulocyte colony-stimulating factor

ROS:

Reactive oxygen species

MCP:

Monocyte chemoattractant proteins

MIP:

Macrophage inflammatory proteins

ECM:

Extracellular matrix

SERPIN-B1:

Serine protease inhibitor

NE:

Neutrophil elastase

MMP-9:

Metalloproteinases-9

YS:

Yolk sac

References

  1. Niedermaier S, Hilgendorff A (2015) Bronchopulmonary dysplasia - an overview about pathophysiologic concepts. Mol Cell Pediatr 2:2. https://doi.org/10.1186/s40348-015-0013-7

    Article  PubMed  PubMed Central  Google Scholar 

  2. Glass HC, Costarino AT, Stayer SA et al (2015) Outcomes for extremely premature infants. Anesth Analg 120:1337–1351. https://doi.org/10.1213/ANE.0000000000000705

    Article  PubMed  PubMed Central  Google Scholar 

  3. McGeachie MJ, Yates KP, Zhou X et al (2016) Patterns of growth and decline in lung function in persistent childhood asthma. N Engl J Med 374:1842–1852. https://doi.org/10.1056/NEJMoa1513737

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Tong Y, Zhang S, Riddle S et al (2021) Intrauterine hypoxia and epigenetic programming in lung development and disease. Biomedicines 9:944. https://doi.org/10.3390/biomedicines9080944

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Biniwale MA, Ehrenkranz RA (2006) The role of nutrition in the prevention and management of bronchopulmonary dysplasia. Semin Perinatol 30:200–208. https://doi.org/10.1053/j.semperi.2006.05.007

    Article  PubMed  Google Scholar 

  6. Trembath A, Laughon MM (2012) Predictors of bronchopulmonary dysplasia. Clin Perinatol 39:585–601. https://doi.org/10.1016/j.clp.2012.06.014

    Article  PubMed  PubMed Central  Google Scholar 

  7. Riyas PK, Vijayakumar KM, Kulkarni ML (2003) Neonatal mechanical ventilation. Indian J Pediatr 70:537–540. https://doi.org/10.1007/BF02723151

    Article  CAS  PubMed  Google Scholar 

  8. Mokres LM, Parai K, Hilgendorff A et al (2010) Prolonged mechanical ventilation with air induces apoptosis and causes failure of alveolar septation and angiogenesis in lungs of newborn mice. Am J Physiol Cell Mol Physiol 298:L23–L35. https://doi.org/10.1152/ajplung.00251.2009

    Article  CAS  Google Scholar 

  9. Jong E, Strunk T, Burgner D et al (2017) The phenotype and function of preterm infant monocytes: implications for susceptibility to infection. J Leukoc Biol 102:645–656. https://doi.org/10.1189/jlb.4ru0317-111r

    Article  CAS  PubMed  Google Scholar 

  10. Driscoll KE, Carter JM, Hassenbein DG, Howard B (1997) Cytokines and particle-induced inflammatory cell recruitment. Environ Health Perspect 105:1159–1164. https://doi.org/10.1289/ehp.97105s51159

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Groneck P, Götze-Speer B, Oppermann M et al (1994) Association of pulmonary inflammation and increased microvascular permeability during the development of bronchopulmonary dysplasia: a sequential analysis of inflammatory mediators in respiratory fluids of high-risk preterm neonates. Pediatrics 93:712–718

    Article  CAS  Google Scholar 

  12. Sun Y, Chen C, Zhang X et al (2019) High neutrophil-to-lymphocyte ratio is an early predictor of bronchopulmonary dysplasia. Front Pediatr 7:1–9. https://doi.org/10.3389/fped.2019.00464

    Article  CAS  Google Scholar 

  13. Kramer BW, Kallapur S, Newnham J, Jobe AH (2009) Prenatal inflammation and lung development. Semin Fetal Neonatal Med 14:2–7. https://doi.org/10.1016/j.siny.2008.08.011

    Article  PubMed  Google Scholar 

  14. Balany J, Bhandari V (2015) Understanding the impact of infection, inflammation, and their persistence in the pathogenesis of bronchopulmonary dysplasia. Front Med 2:1–10. https://doi.org/10.3389/fmed.2015.00090

    Article  Google Scholar 

  15. Wright RJ, Visness CM, Calatroni A et al (2010) Prenatal maternal stress and cord blood innate and adaptive cytokine responses in an inner-city cohort. Am J Respir Crit Care Med 182:25–33. https://doi.org/10.1164/rccm.200904-0637OC

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Humberg A, Fortmann I, Siller B et al (2020) Preterm birth and sustained inflammation: consequences for the neonate. Semin Immunopathol 42:451–468. https://doi.org/10.1007/s00281-020-00803-2

    Article  PubMed  PubMed Central  Google Scholar 

  17. Agrawal V, Hirsch E (2012) Intrauterine infection and preterm labor. Semin Fetal Neonatal Med 17:12–19. https://doi.org/10.1016/j.siny.2011.09.001

    Article  PubMed  Google Scholar 

  18. Basha S, Surendran N, Pichichero M (2014) Immune responses in neonates. Expert Rev Clin Immunol 10:1171–1184. https://doi.org/10.1586/1744666X.2014.942288

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Palmeira P, Quinello C, Silveira-Lessa AL et al (2012) IgG placental transfer in healthy and pathological pregnancies. Clin Dev Immunol 2012:1–13. https://doi.org/10.1155/2012/985646

    Article  CAS  Google Scholar 

  20. Speer CP (2006) Pulmonary inflammation and bronchopulmonary dysplasia. J Perinatol 26:S57–S62. https://doi.org/10.1038/sj.jp.7211476

    Article  CAS  PubMed  Google Scholar 

  21. Speer CP (2006) Inflammation and bronchopulmonary dysplasia: a continuing story. Semin Fetal Neonatal Med 11:354–362

    Article  Google Scholar 

  22. Goetz MJ, Kremer S, Behnke J et al (2021) MSC based therapies to prevent or treat BPD—a narrative review on advances and ongoing challenges. Int J Mol Sci 22:1138. https://doi.org/10.3390/ijms22031138

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Heydarian M, Oak P, Zhang X et al (2022) Relationship between impaired BMP signalling and clinical risk factors at early-stage vascular injury in the preterm infant. Thorax thoraxjnl-2021-218083. https://doi.org/10.1136/thoraxjnl-2021-218083

  24. Eldredge LC, Creasy RS, Presnell S et al (2019) Infants with evolving bronchopulmonary dysplasia demonstrate monocyte-specific expression of IL-1 in tracheal aspirates. Am J Physiol Cell Mol Physiol 317:L49–L56. https://doi.org/10.1152/ajplung.00060.2019

    Article  CAS  Google Scholar 

  25. Levy O (2007) Innate immunity of the newborn: basic mechanisms and clinical correlates. Nat Rev Immunol 7:379–390. https://doi.org/10.1038/nri2075

    Article  CAS  PubMed  Google Scholar 

  26. Yu JC, Khodadadi H, Malik A et al (2018) Innate immunity of neonates and infants. Front Immunol 9. https://doi.org/10.3389/fimmu.2018.01759

  27. Newburg DS, Walker WA (2007) Protection of the neonate by the innate immune system of developing gut and of human milk. Pediatr Res 61:2–8. https://doi.org/10.1203/01.pdr.0000250274.68571.18

    Article  CAS  PubMed  Google Scholar 

  28. MacGillivray DM, Kollmann TR (2014) The role of environmental factors in modulating immune responses in early life. Front Immunol 5:ji2101192. https://doi.org/10.3389/fimmu.2014.00434

    Article  CAS  Google Scholar 

  29. Sokol CL, Luster AD (2015) The chemokine system in innate immunity. Cold Spring Harb Perspect Biol 7:a016303. https://doi.org/10.1101/cshperspect.a016303

    Article  PubMed  PubMed Central  Google Scholar 

  30. Clark R, Kupper T (2005) Old meets new: the interaction between innate and adaptive immunity. J Invest Dermatol 125:629–637. https://doi.org/10.1111/j.0022-202X.2005.23856.x

    Article  CAS  PubMed  Google Scholar 

  31. Arango Duque G, Descoteaux A (2014) Macrophage cytokines: involvement in immunity and infectious diseases. Front Immunol 5:1–12. https://doi.org/10.3389/fimmu.2014.00491

    Article  CAS  Google Scholar 

  32. Ibrahim J, Garantziotis S, Savani RC (2020) The inflammation superhighway. In: Updates on neonatal chronic lung disease. Elsevier, Philadelphia, pp 131–150. https://doi.org/10.1016/B978-0-323-68353-1.00009-9

  33. Martin TR (2005) Innate immunity in the lungs. Proc Am Thorac Soc 2:403–411. https://doi.org/10.1513/pats.200508-090JS

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Glaser, Coulter, Shields et al (2019) Airway epithelial derived cytokines and chemokines and their role in the immune response to respiratory syncytial virus infection. Pathogens 8:106. https://doi.org/10.3390/pathogens8030106

    Article  CAS  PubMed Central  Google Scholar 

  35. Hewitt RJ, Lloyd CM (2021) Regulation of immune responses by the airway epithelial cell landscape. Nat Rev Immunol 21:347–362. https://doi.org/10.1038/s41577-020-00477-9

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Qing C, Ziyun L, Xuefei Y et al (2022) Protective effects of 18β-glycyrrhetinic acid on neonatal rats with hyperoxia exposure. Inflammation. https://doi.org/10.1007/s10753-021-01616-7

  37. Heydarian M, Schweinlin M, Schwarz T et al (2021) Triple co-culture and perfusion bioreactor for studying the interaction between Neisseria gonorrhoeae and neutrophils: a novel 3D tissue model for bacterial infection and immunity. J Tissue Eng 12:204173142098880. https://doi.org/10.1177/2041731420988802

    Article  CAS  Google Scholar 

  38. Misharin AV, Morales-Nebreda L, Reyfman PA et al (2017) Monocyte-derived alveolar macrophages drive lung fibrosis and persist in the lung over the life span. J Exp Med 214:2387–2404. https://doi.org/10.1084/jem.20162152

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Italiani P, Boraschi D (2014) From monocytes to M1/M2 macrophages: phenotypical vs. functional differentiation. Front Immunol 5:1–22. https://doi.org/10.3389/fimmu.2014.00514

    Article  CAS  Google Scholar 

  40. Honda A, Hoeksema MA, Sakai M et al (2022) The lung microenvironment instructs gene transcription in neonatal and adult alveolar macrophages. J Immunol 26:ji2101192. https://doi.org/10.4049/jimmunol.2101192

    Article  CAS  Google Scholar 

  41. Ambalavanan N, Carlo WA, D’Angio CT et al (2009) Cytokines associated with bronchopulmonary dysplasia or death in extremely low birth weight infants. Pediatrics 123:1132–1141. https://doi.org/10.1542/peds.2008-0526

    Article  PubMed  Google Scholar 

  42. Baier RJ, Majid A, Parupia H et al (2004) CC chemokine concentrations increase in respiratory distress syndrome and correlate with development of bronchopulmonary dysplasia. Pediatr Pulmonol 37:137–148. https://doi.org/10.1002/ppul.10417

    Article  PubMed  Google Scholar 

  43. Bose CL, Dammann CEL, Laughon MM (2008) Bronchopulmonary dysplasia and inflammatory biomarkers in the premature neonate. Arch Dis Child - Fetal Neonatal Ed 93:F455–F461. https://doi.org/10.1136/adc.2007.121327

    Article  CAS  PubMed  Google Scholar 

  44. Wagenaar GTM, ter Horst SA, van Gastelen MA et al (2004) Gene expression profile and histopathology of experimental bronchopulmonary dysplasia induced by prolonged oxidative stress. Free Radic Biol Med 36:782–801. https://doi.org/10.1016/j.freeradbiomed.2003.12.007

    Article  CAS  PubMed  Google Scholar 

  45. Köksal N, Kayık B, Çetinkaya M et al (2012) Value of serum and bronchoalveolar fluid lavage pro- and anti-inflammatory cytokine levels for predicting bronchopulmonary dysplasia in premature infants. Eur Cytokine Netw 23:29–35. https://doi.org/10.1684/ecn.2012.0304

    Article  CAS  PubMed  Google Scholar 

  46. Baier RJ, Loggins J, Kruger TE (2003) Interleukin-4 and 13 concentrations in infants at risk to develop Bronchopulmonary Dysplasia. BMC Pediatr 3:8. https://doi.org/10.1186/1471-2431-3-8

    Article  PubMed  PubMed Central  Google Scholar 

  47. Cai J, Lu H, Su Z et al (2022) Dynamic changes of NCR− type 3 innate lymphoid cells and their role in mice with bronchopulmonary dysplasia. Inflammation. https://doi.org/10.1007/s10753-021-01543-7

  48. Kuwabara T, Ishikawa F, Kondo M, Kakiuchi T (2017) The role of IL-17 and related cytokines in inflammatory autoimmune diseases. Mediators Inflamm 2017:1–11. https://doi.org/10.1155/2017/3908061

    Article  CAS  Google Scholar 

  49. Schibler KR, Liechty KW, White WL, Christensen RD (1993) Production of granulocyte colony-stimulating factor in vitro by monocytes from preterm and term neonates. Blood 82:2478–2484. https://doi.org/10.1182/blood.V82.8.2478.2478

    Article  CAS  PubMed  Google Scholar 

  50. Wright CJ, Kirpalani H (2011) Targeting inflammation to prevent bronchopulmonary dysplasia: can new insights be translated into therapies? Pediatrics 128:111–126. https://doi.org/10.1542/peds.2010-3875

    Article  PubMed  PubMed Central  Google Scholar 

  51. Savani RC (2018) Modulators of inflammation in bronchopulmonary dysplasia. Semin Perinatol 42:459–470. https://doi.org/10.1053/j.semperi.2018.09.009

    Article  PubMed  PubMed Central  Google Scholar 

  52. Beck-Schimmer B, Schwendener R, Pasch T et al (2005) Alveolar macrophages regulate neutrophil recruitment in endotoxin-induced lung injury. Respir Res 6:61. https://doi.org/10.1186/1465-9921-6-61

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Phillipson M, Kubes P (2011) The neutrophil in vascular inflammation. Nat Med 17:1381–1390. https://doi.org/10.1038/nm.2514

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Aggarwal NR, King LS, D’Alessio FR (2014) Diverse macrophage populations mediate acute lung inflammation and resolution. Am J Physiol Cell Mol Physiol 306:L709–L725. https://doi.org/10.1152/ajplung.00341.2013

    Article  CAS  Google Scholar 

  55. Christensen RD (1989) Hematopoiesis in the fetus and neonate. Pediatr Res 26:531–532. https://doi.org/10.1203/00006450-198912000-00001

    Article  CAS  PubMed  Google Scholar 

  56. Furze RC, Rankin SM (2008) Neutrophil mobilization and clearance in the bone marrow. Immunology 125:281–288. https://doi.org/10.1111/j.1365-2567.2008.02950.x

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Prame Kumar K, Nicholls AJ, Wong CHY (2018) Partners in crime: neutrophils and monocytes/macrophages in inflammation and disease. Cell Tissue Res 371:551–565. https://doi.org/10.1007/s00441-017-2753-2

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Nauseef WM, Borregaard N (2014) Neutrophils at work. Nat Immunol 15:602–611. https://doi.org/10.1038/ni.2921

    Article  CAS  PubMed  Google Scholar 

  59. Bayr H (2005) Reactive oxygen species. Crit Care Med 33:S498–S501. https://doi.org/10.1097/01.CCM.0000186787.64500.12

    Article  Google Scholar 

  60. Wen ST, Chen W, Chen HL et al (2013) Amniotic fluid stem cells from EGFP transgenic mice attenuate hyperoxia-induced acute lung injury. PLoS One 8. https://doi.org/10.1371/journal.pone.0075383

  61. Turunen R, Nupponen I, Siitonen S et al (2006) Onset of mechanical ventilation is associated with rapid activation of circulating phagocytes in preterm infants. Pediatrics 117:448–454. https://doi.org/10.1542/peds.2005-0123

    Article  PubMed  Google Scholar 

  62. Yildiz C, Palaniyar N, Otulakowski G et al (2015) Mechanical ventilation induces neutrophil extracellular trap formation. Anesthesiology 122:864–875. https://doi.org/10.1097/ALN.0000000000000605

    Article  CAS  PubMed  Google Scholar 

  63. Thomas JM, Sudhadevi T, Basa P et al (2022) The role of sphingolipid signaling in oxidative lung injury and pathogenesis of bronchopulmonary dysplasia. Int J Mol Sci 23:1254. https://doi.org/10.3390/ijms23031254

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Wang J, Dong W (2018) Oxidative stress and bronchopulmonary dysplasia. Gene 678:177–183. https://doi.org/10.1016/j.gene.2018.08.031

    Article  CAS  PubMed  Google Scholar 

  65. Vargas A, Roux-Dalvai F, Droit A, Lavoie J-P (2016) Neutrophil-derived exosomes: a new mechanism contributing to airway smooth muscle remodeling. Am J Respir Cell Mol Biol 55:450–461. https://doi.org/10.1165/rcmb.2016-0033OC

    Article  CAS  PubMed  Google Scholar 

  66. Genschmer KR, Russell DW, Lal C et al (2019) Activated PMN exosomes: pathogenic entities causing matrix destruction and disease in the lung. Cell 176:113–126.e15. https://doi.org/10.1016/j.cell.2018.12.002

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Yi M, Jankov RP, Belcastro R et al (2004) Opposing effects of 60% oxygen and neutrophil influx on alveologenesis in the neonatal rat. Am J Respir Crit Care Med 170:1188–1196. https://doi.org/10.1164/rccm.200402-215OC

    Article  PubMed  Google Scholar 

  68. Mižíková I, Morty RE (2015) The extracellular matrix in bronchopulmonary dysplasia: target and source. Front Med 2:1–20. https://doi.org/10.3389/fmed.2015.00091

    Article  Google Scholar 

  69. Xu J, Mao X, Jin R et al (2020) Neutrophil extracellular traps degrade fibronectin in a rat model of bronchopulmonary dysplasia induced by perinatal exposure to lipopolysaccharide. J Cell Mol Med 24:14645–14649. https://doi.org/10.1111/jcmm.15842

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Zhu Y, Huang Y, Ji Q et al (2021) Interplay between extracellular matrix and neutrophils in diseases. J Immunol Res 2021:1–11. https://doi.org/10.1155/2021/8243378

    Article  CAS  Google Scholar 

  71. Yasumatsu R, Altiok O, Benarafa C et al (2006) SERPINB1 upregulation is associated with in vivo complex formation with neutrophil elastase and cathepsin G in a baboon model of bronchopulmonary dysplasia. Am J Physiol Cell Mol Physiol 291:L619–L627. https://doi.org/10.1152/ajplung.00507.2005

    Article  CAS  Google Scholar 

  72. Carr R (2000) Neutrophil production and function in newborn infants. Br J Haematol 110:18–28. https://doi.org/10.1046/j.1365-2141.2000.01992.x

    Article  CAS  PubMed  Google Scholar 

  73. Hellebrekers P, Vrisekoop N, Koenderman L (2018) Neutrophil phenotypes in health and disease. Eur J Clin Invest 48:e12943. https://doi.org/10.1111/eci.12943

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Yost CC, Cody MJ, Harris ES et al (2009) Impaired neutrophil extracellular trap (NET) formation: a novel innate immune deficiency of human neonates. Blood 113:6419–6427. https://doi.org/10.1182/blood-2008-07-171629

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Amulic B, Cazalet C, Hayes GL et al (2012) Neutrophil function: from mechanisms to disease. Annu Rev Immunol 30:459–489. https://doi.org/10.1146/annurev-immunol-020711-074942

    Article  CAS  PubMed  Google Scholar 

  76. Ballabh P (2004) Neutrophil and monocyte adhesion molecules in bronchopulmonary dysplasia, and effects of corticosteroids. Arch Dis Child - Fetal Neonatal Ed 89:76–83. https://doi.org/10.1136/fn.89.1.F76

    Article  Google Scholar 

  77. Li X, Wang Q, Luo T, Li T (2020) Decreased neutrophil levels in bronchopulmonary dysplasia infants. Pediatr Neonatol 61:637–644. https://doi.org/10.1016/j.pedneo.2020.08.013

    Article  PubMed  Google Scholar 

  78. Kroon AA, Wang J, Huang Z et al (2010) Inflammatory response to oxygen and endotoxin in newborn rat lung ventilated with low tidal volume. Pediatr Res 68:63–69. https://doi.org/10.1203/PDR.0b013e3181e17caa

    Article  CAS  PubMed  Google Scholar 

  79. Grigg JM, Silverman M, Savill JS et al (1991) Neutrophil apoptosis and clearance from neonatal lungs. Lancet 338:720–722. https://doi.org/10.1016/0140-6736(91)91443-X

    Article  CAS  PubMed  Google Scholar 

  80. Scott CL, Henri S, Guilliams M (2014) Mononuclear phagocytes of the intestine, the skin, and the lung. Immunol Rev 262:9–24. https://doi.org/10.1111/imr.12220

    Article  CAS  PubMed  Google Scholar 

  81. Landsman L, Jung S (2007) Lung macrophages serve as obligatory intermediate between blood monocytes and alveolar macrophages. J Immunol 179:3488–3494. https://doi.org/10.4049/jimmunol.179.6.3488

    Article  CAS  PubMed  Google Scholar 

  82. McQuattie-Pimentel AC, Budinger GRS, Ballinger MN (2018) Monocyte-derived alveolar macrophages: the dark side of lung repair? Am J Respir Cell Mol Biol 58:5–6. https://doi.org/10.1165/rcmb.2017-0328ED

    Article  CAS  PubMed  Google Scholar 

  83. Kalymbetova TV, Selvakumar B, Rodríguez-Castillo JA et al (2018) Resident alveolar macrophages are master regulators of arrested alveolarization in experimental bronchopulmonary dysplasia. J Pathol 245:153–159. https://doi.org/10.1002/path.5076

    Article  CAS  PubMed  Google Scholar 

  84. Sanchez-Schmitz G, Morrocchi E, Cooney M et al (2020) Neonatal monocytes demonstrate impaired homeostatic extravasation into a microphysiological human vascular model. Sci Rep 10:17836. https://doi.org/10.1038/s41598-020-74639-z

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Shi C, Pamer EG (2011) Monocyte recruitment during infection and inflammation. Nat Rev Immunol 11:762–774. https://doi.org/10.1038/nri3070

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Gerhardt T, Ley K (2015) Monocyte trafficking across the vessel wall. Cardiovasc Res 107:321–330. https://doi.org/10.1093/cvr/cvv147

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Lechner AJ, Driver IH, Lee J et al (2017) Recruited monocytes and type 2 immunity promote lung regeneration following pneumonectomy. Cell Stem Cell 21:120–134.e7. https://doi.org/10.1016/j.stem.2017.03.024

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Hallwirth U, Pomberger G, Pollak A et al (2004) Monocyte switch in neonates: high phagocytic capacity and low HLA-DR expression in VLBWI are inverted during gestational aging. Pediatr Allergy Immunol 15:513–516. https://doi.org/10.1111/j.1399-3038.2004.00168.x

    Article  PubMed  Google Scholar 

  89. Kapellos TS, Bonaguro L, Gemünd I et al (2019) Human monocyte subsets and phenotypes in major chronic inflammatory diseases. Front Immunol 10:1–13. https://doi.org/10.3389/fimmu.2019.02035

    Article  CAS  Google Scholar 

  90. Ong S-M, Teng K, Newell E et al (2019) A novel, five-marker alternative to CD16–CD14 gating to identify the three human monocyte subsets. Front Immunol 10:1761. https://doi.org/10.3389/fimmu.2019.01761

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Chimen M, Yates CM, McGettrick HM et al (2017) Monocyte subsets coregulate inflammatory responses by integrated signaling through TNF and IL-6 at the endothelial cell interface. J Immunol 198:2834–2843. https://doi.org/10.4049/jimmunol.1601281

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Narasimhan PB, Marcovecchio P, Hamers AAJ, Hedrick CC (2019) Nonclassical monocytes in health and disease. Annu Rev Immunol 37:439–456. https://doi.org/10.1146/annurev-immunol-042617-053119

    Article  CAS  PubMed  Google Scholar 

  93. Spahn JH, Kreisel D (2014) Monocytes in sterile inflammation: recruitment and functional consequences. Arch Immunol Ther Exp (Warsz) 62:187–194. https://doi.org/10.1007/s00005-013-0267-5

    Article  CAS  Google Scholar 

  94. Geissmann F, Jung S, Littman DR (2003) Blood monocytes consist of two principal subsets with distinct migratory properties. Immunity 19:71–82. https://doi.org/10.1016/S1074-7613(03)00174-2

    Article  CAS  PubMed  Google Scholar 

  95. Ziegler-Heitbrock L, Ancuta P, Crowe S et al (2010) Nomenclature of monocytes and dendritic cells in blood. Blood 116:74–80. https://doi.org/10.1182/blood-2010-02-258558

    Article  CAS  Google Scholar 

  96. Carlin LM, Stamatiades EG, Auffray C et al (2013) Nr4a1-dependent Ly6Clow monocytes monitor endothelial cells and orchestrate their disposal. Cell 153:362–375. https://doi.org/10.1016/j.cell.2013.03.010

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Wynn TA, Chawla A, Pollard JW (2013) Macrophage biology in development, homeostasis and disease. Nature 496:445–455. https://doi.org/10.1038/nature12034

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Glod J, Kobiler D, Noel M et al (2006) Monocytes form a vascular barrier and participate in vessel repair after brain injury. Blood 107:940–946. https://doi.org/10.1182/blood-2004-11-4403

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Hernandez GE, Iruela-Arispe ML (2020) The many flavors of monocyte/macrophage--endothelial cell interactions. Curr Opin Hematol 27:181–189. https://doi.org/10.1097/MOH.0000000000000573

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. van Gils JM, Zwaginga JJ, Hordijk PL (2009) Molecular and functional interactions among monocytes, platelets, and endothelial cells and their relevance for cardiovascular diseases. J Leukoc Biol 85:195–204. https://doi.org/10.1189/jlb.0708400

    Article  CAS  PubMed  Google Scholar 

  101. Hakkert B, Kuijpers T, Leeuwenberg J et al (1991) Neutrophil and monocyte adherence to and migration across monolayers of cytokine-activated endothelial cells: the contribution of CD18, ELAM-1, and VLA-4. Blood 78:2721–2726. https://doi.org/10.1182/blood.V78.10.2721.2721

    Article  CAS  PubMed  Google Scholar 

  102. Gordon S (1995) The macrophage. BioEssays 17:977–986. https://doi.org/10.1002/bies.950171111

    Article  CAS  PubMed  Google Scholar 

  103. Kaufman D, Kilpatrick L, Hudson RG et al (1999) Decreased superoxide production, degranulation, tumor necrosis factor alpha secretion, and CD11b/CD18 receptor expression by adherent monocytes from preterm infants. Clin Diagnostic Lab Immunol 6:525–529. https://doi.org/10.1128/CDLI.6.4.525-529.1999

    Article  CAS  Google Scholar 

  104. Oak P, Hilgendorff A (2017) The BPD trio? Interaction of dysregulated PDGF, VEGF, and TGF signaling in neonatal chronic lung disease. Mol Cell Pediatr 4. https://doi.org/10.1186/s40348-017-0076-8

  105. Bartram U, Speer CP (2004) The role of transforming growth factor β in lung development and disease. Chest 125:754–765. https://doi.org/10.1378/chest.125.2.754

    Article  PubMed  Google Scholar 

  106. Alejandre-Alcázar MA, Michiels-Corsten M, Vicencio AG et al (2008) TGF-β signaling is dynamically regulated during the alveolarization of rodent and human lungs. Dev Dyn 237:259–269. https://doi.org/10.1002/dvdy.21403

    Article  CAS  PubMed  Google Scholar 

  107. Kunzmann S, Speer CP, Jobe AH, Kramer BW (2007) Antenatal inflammation induced TGF-β1 but suppressed CTGF in preterm lungs. Am J Physiol Cell Mol Physiol 292:L223–L231. https://doi.org/10.1152/ajplung.00159.2006

    Article  CAS  Google Scholar 

  108. Yu X, Buttgereit A, Lelios I et al (2017) The cytokine TGF-β promotes the development and homeostasis of alveolar macrophages. Immunity 47:903–912.e4. https://doi.org/10.1016/j.immuni.2017.10.007

    Article  CAS  PubMed  Google Scholar 

  109. Wahl SM, Hunt DA, Wakefield LM et al (1987) Transforming growth factor type beta induces monocyte chemotaxis and growth factor production. Proc Natl Acad Sci 84:5788–5792. https://doi.org/10.1073/pnas.84.16.5788

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Kelly A, Gunaltay S, McEntee CP et al (2018) Human monocytes and macrophages regulate immune tolerance via integrin αvβ8–mediated TGFβ activation. J Exp Med 215:2725–2736. https://doi.org/10.1084/jem.20171491

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  111. Lavin Y, Winter D, Blecher-Gonen R et al (2014) Tissue-resident macrophage enhancer landscapes are shaped by the local microenvironment. Cell 159:1312–1326. https://doi.org/10.1016/j.cell.2014.11.018

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Perdiguero EG, Klapproth K, Schulz C et al (2015) Tissue-resident macrophages originate from yolk sac-derived erythro-myeloid progenitors. Exp Hematol 43:S64. https://doi.org/10.1016/j.exphem.2015.06.130

    Article  Google Scholar 

  113. Hoeffel G, Chen J, Lavin Y et al (2015) C-Myb+ erythro-myeloid progenitor-derived fetal monocytes give rise to adult tissue-resident macrophages. Immunity 42:665–678. https://doi.org/10.1016/j.immuni.2015.03.011

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  114. Kopf M, Schneider C, Nobs SP (2015) The development and function of lung-resident macrophages and dendritic cells. Nat Immunol 16:36–44. https://doi.org/10.1038/ni.3052

    Article  CAS  PubMed  Google Scholar 

  115. Guilliams M, De Kleer I, Henri S et al (2013) Alveolar macrophages develop from fetal monocytes that differentiate into long-lived cells in the first week of life via GM-CSF. J Exp Med 210:1977–1992. https://doi.org/10.1084/jem.20131199

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. van de Laar L, Saelens W, De Prijck S et al (2016) Yolk sac macrophages, fetal liver, and adult monocytes can colonize an empty niche and develop into functional tissue-resident macrophages. Immunity 44:755–768. https://doi.org/10.1016/j.immuni.2016.02.017

    Article  CAS  PubMed  Google Scholar 

  117. Evren E, Ringqvist E, Doisne J-M et al (2022) CD116+ fetal precursors migrate to the perinatal lung and give rise to human alveolar macrophages. J Exp Med 219. https://doi.org/10.1084/jem.20210987

  118. Yang J, Zhang L, Yu C et al (2014) Monocyte and macrophage differentiation: circulation inflammatory monocyte as biomarker for inflammatory diseases. Biomark Res 2:1–9. https://doi.org/10.1186/2050-7771-2-1

    Article  PubMed  PubMed Central  Google Scholar 

  119. Milan A, Priante E, Nardo D et al (2017) Early macrophage activation in preterm newborns and respiratory disease. J Child Sci 07:e110–e119. https://doi.org/10.1055/s-0037-1605579

    Article  Google Scholar 

  120. Atochina-Vasserman EN, Bates SR, Zhang P et al (2011) Early alveolar epithelial dysfunction promotes lung inflammation in a mouse model of Hermansky-Pudlak Syndrome. Am J Respir Crit Care Med 184:449–458. https://doi.org/10.1164/rccm.201011-1882OC

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  121. Gibbons MA, MacKinnon AC, Ramachandran P et al (2011) Ly6C hi monocytes direct alternatively activated profibrotic macrophage regulation of lung fibrosis. Am J Respir Crit Care Med 184:569–581. https://doi.org/10.1164/rccm.201010-1719OC

    Article  CAS  PubMed  Google Scholar 

  122. Janssen WJ, Barthel L, Muldrow A et al (2011) Fas determines differential fates of resident and recruited macrophages during resolution of acute lung injury. Am J Respir Crit Care Med 184:547–560. https://doi.org/10.1164/rccm.201011-1891OC

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  123. Sahoo D, Zaramela LS, Hernandez GE et al (2020) Transcriptional profiling of lung macrophages identifies a predictive signature for inflammatory lung disease in preterm infants. Commun Biol 3:259. https://doi.org/10.1038/s42003-020-0985-2

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. Kramer BW, Jobe AH, Ikegami M (2003) Monocyte function in preterm, term, and adult sheep. Pediatr Res 54:52–57. https://doi.org/10.1203/01.PDR.0000066621.11877.33

    Article  PubMed  Google Scholar 

  125. Dreschers S, Ohl K, Schulte N et al (2020) Impaired functional capacity of polarised neonatal macrophages. Sci Rep 10:1–12. https://doi.org/10.1038/s41598-019-56928-4

    Article  CAS  Google Scholar 

  126. Hegge I, Niepel F, Lange A et al (2019) Functional analysis of granulocyte and monocyte subpopulations in neonates. Mol Cell Pediatr 6. https://doi.org/10.1186/s40348-019-0092-y

  127. Yachie A, Takano N, Ohta K et al (1992) Defective production of interleukin-6 in very small premature infants in response to bacterial pathogens. Infect Immun 60:749–753. https://doi.org/10.1128/iai.60.3.749-753.1992

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. Currie AJ, Curtis S, Strunk T et al (2011) Preterm infants have deficient monocyte and lymphocyte cytokine responses to group B Streptococcus. Infect Immun 79:1588–1596. https://doi.org/10.1128/IAI.00535-10

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. Davidson L, Berkelhamer S (2017) Bronchopulmonary dysplasia: chronic lung disease of infancy and long-term pulmonary outcomes. J Clin Med 6:4. https://doi.org/10.3390/jcm6010004

    Article  CAS  PubMed Central  Google Scholar 

  130. Tullus K, Noack GW, Burman LG et al (1996) Elevated cytokine levels in tracheobronchial aspirate fluids from ventilator treated neonates with bronchopulmonary dysplasia. Eur J Pediatr 155:112–116. https://doi.org/10.1007/BF02075762

    Article  CAS  PubMed  Google Scholar 

  131. Pugin J, Dunn I, Jolliet P et al (1998) Activation of human macrophages by mechanical ventilation in vitro. Am J Physiol 275:L1040–L1050. https://doi.org/10.1152/ajplung.1998.275.6.L1040

    Article  CAS  PubMed  Google Scholar 

  132. Jonsson B, Tullus K, Brauner A et al (1997) Early increase of TNFalpha and IL-6 in tracheobronchial aspirate fluid indicator of subsequent chronic lung disease in preterm infants. Arch Dis Child - Fetal Neonatal Ed 77:F198–F201. https://doi.org/10.1136/fn.77.3.F198

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  133. Li L, Chen J, Xiong G et al (2016) Increased ROS production in non-polarized mammary epithelial cells induces monocyte infiltration in 3D culture. J Cell Sci 130:190–202. https://doi.org/10.1242/jcs.186031

    Article  CAS  PubMed  Google Scholar 

  134. Ogden BE, Murphy SA, Saunders GC et al (1984) Neonatal lung neutrophils and elastase/proteinase inhibitor imbalance. Am Rev Respir Dis 130:817–821. https://doi.org/10.1164/arrd.1984.130.5.817

    Article  CAS  PubMed  Google Scholar 

  135. Baier JR, Loggins J, Kruger TE (2001) Monocyte chemoattractant protein-1 and interleukin-8 are increased in bronchopulmonary dysplasia. J Investig Med 49:362–369. https://doi.org/10.2310/6650.2001.33902

    Article  CAS  PubMed  Google Scholar 

  136. Davidson D, Miskolci V, Clark DC et al (2007) Interleukin-10 production after pro-inflammatory stimulation of neutrophils and monocytic cells of the newborn. Neonatology 92:127–133. https://doi.org/10.1159/000101432

    Article  CAS  PubMed  Google Scholar 

  137. Nielsen MC, Andersen MN, Møller HJ (2020) Monocyte isolation techniques significantly impact the phenotype of both isolated monocytes and derived macrophages in vitro. Immunology 159:63–74. https://doi.org/10.1111/imm.13125

    Article  CAS  PubMed  Google Scholar 

  138. Ehrhardt H, Pritzke T, Oak P et al (2016) Absence of TNF-α enhances inflammatory response in the newborn lung undergoing mechanical ventilation. Am J Physiol - Lung Cell Mol Physiol 310:L909–L918. https://doi.org/10.1152/ajplung.00367.2015

    Article  PubMed  PubMed Central  Google Scholar 

  139. Oak P, Pritzke T, Thiel I et al (2017) Attenuated PDGF signaling drives alveolar and microvascular defects in neonatal chronic lung disease. EMBO Mol Med 9:1504–1520. https://doi.org/10.15252/emmm.201607308

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  140. Pruenster M, Kurz ARM, Chung K-J et al (2015) Extracellular MRP8/14 is a regulator of β2 integrin-dependent neutrophil slow rolling and adhesion. Nat Commun 6:6915. https://doi.org/10.1038/ncomms7915

    Article  CAS  PubMed  Google Scholar 

  141. Doryab A, Tas S, Taskin MB et al (2019) Evolution of bioengineered lung models: recent advances and challenges in tissue mimicry for studying the role of mechanical forces in cell biology. Adv Funct Mater 29:1903114. https://doi.org/10.1002/adfm.201903114

    Article  CAS  Google Scholar 

  142. Palta M, Sadek-Badawi M, Carlton DP (2008) Association of BPD and IVH with early neutrophil and white counts in VLBW neonates with gestational age <32 weeks. J Perinatol 28:604–610. https://doi.org/10.1038/jp.2008.65

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  143. Hilgendorff A, Parai K, Ertsey R et al (2011) Inhibiting lung elastase activity enables lung growth in mechanically ventilated newborn mice. Am J Respir Crit Care Med 184:537–546. https://doi.org/10.1164/rccm.201012-2010OC

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  144. Nold MF, Mangan NE, Rudloff I et al (2013) Interleukin-1 receptor antagonist prevents murine bronchopulmonary dysplasia induced by perinatal inflammation and hyperoxia. Proc Natl Acad Sci 110:14384–14389. https://doi.org/10.1073/pnas.1306859110

    Article  PubMed  PubMed Central  Google Scholar 

  145. Sureshbabu A, Syed MA, Boddupalli CS et al (2015) Conditional overexpression of TGFβ1 promotes pulmonary inflammation, apoptosis and mortality via TGFβR2 in the developing mouse lung. Respir Res 16:4. https://doi.org/10.1186/s12931-014-0162-6

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  146. Alam MA, Betal SGN, Aghai ZH, Bhandari V (2019) Hyperoxia causes miR199a-5p-mediated injury in the developing lung. Pediatr Res 86:579–588. https://doi.org/10.1038/s41390-019-0524-3

    Article  CAS  PubMed  Google Scholar 

  147. Shrestha D, Ye GX, Stabley D et al (2021) Pulmonary immune cell transcriptome changes in double-hit model of BPD induced by chorioamnionitis and postnatal hyperoxia. Pediatr Res. https://doi.org/10.1038/s41390-020-01319-z

Download references

Acknowledgements

We strived to cover all the papers related to the topic of the present review paper. We, however, apologize for any missing references. In part, the schematic was created using, the Motifolio biology toolkit.

Funding

Open Access funding enabled and organized by Projekt DEAL. The present study work was supported by the German Center for Lung Research (DZL, German Ministry of Education and Health (BMBF)) as well as the Research Training Group Targets in Toxicology (GRK2338) of the German Science and Research Organization (DFG). Additional support was provided by the Stiftung AtemWeg (LSS AIRR).

Author information

Authors and Affiliations

Authors

Contributions

M. H. and A. H. conceptualized and wrote the first draft of the manuscript. M.H. designed the figure and table. M.H., C. S., T. S., and A. H. wrote and revised the final manuscript. The author(s) read and approved the final manuscript.

Corresponding author

Correspondence to Anne Hilgendorff.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Heydarian, M., Schulz, C., Stoeger, T. et al. Association of immune cell recruitment and BPD development. Mol Cell Pediatr 9, 16 (2022). https://doi.org/10.1186/s40348-022-00148-w

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s40348-022-00148-w

Keywords